Elsevier

Neuroscience Letters

Volume 771, 6 February 2022, 136413
Neuroscience Letters

Fingolimod protects against cerebral ischemia reperfusion injury in rats by reducing inflammatory cytokines and inhibiting the activation of p38 MAPK and NF-κB signaling pathways

https://doi.org/10.1016/j.neulet.2021.136413Get rights and content

Highlights

  • FTY720 attenuates the expression of inflammatory factors IL-1β, IL-6 and TNF-α.

  • FTY720 inhibits the activation of P38 MAPK/NF-κB signaling pathway.

  • FTY720 exhibits neuroprotective effects against ischemic reperfusion injury in rats.

Abstract

Fingolimod (FTY720) is a sphingosine 1-phosphate (S1P) receptor agonist. Here, to understand biological activity of FTY720 pretreatment and post-treatment on cerebral ischemia reperfusion injury (CIRI), rat transient middle cerebral artery occlusion/reperfusion (tMCAO/R) model was generated. Neurological deficit scoring was assessed after tMCAO/R. Four groups were established including sham-operated control group, operated group, and two FTY720-treated groups. Neuron damage was observed by Nissl staining. Gene expression was measured using qPCR and western blot analysis. Tumor necrosis factor α (TNF-α), interleukin 1β (IL-1β) and interleukin 6 (IL-6) levels were evaluated by enzyme-linked immunosorbent assay (ELISA). We uncovered that neurological score in two FTY720-treated groups was significantly lower than that in the operated group. FTY720 pretreatment or posttreatment groups had a significantly increased number of Nissl bodies in cerebral cortex as compared with the operated group, indicating that FTY720 administration reduced neuronal damage. Besides, FTY720 posttreatment improved memory impairment induced by tMCAO/R. In addition, IL-1β, IL-6, and TNF-α levels in the cerebral cortex and hippocampus of two FTY720-treated groups were significantly decreased in comparison to the operated group, showing that FTY720 could reduce the release of inflammatory cytokines in brain tissue. Furthermore, phosphorylation of p38MAPK and NF-κB pathway-related molecules in ischemic brain tissues of FTY720 group were markedly down-regulated compared to the operated group. Together, FTY720 pretreatment or posttreatment improved the neurological deficit of middle cerebral ischemia/reperfusion rat model and reduced neuronal damage by decreasing the levels of inflammatory cytokines and attenuating the phosphorylation levels of p38MAPK and NF-κB pathway-associated molecules. FTY720 exhibits neuroprotective effects against ischemic reperfusion injury in rats.

Introduction

Cerebral ischemia–reperfusion injury is a therapeutic challenge for treatment of ischemic brain damage [33] and reperfusion exacerbates cerebral injury causing brain cell death. Drugs for cerebral ischemia–reperfusion injury have been attracted increasing attention in recent years. Fingolimod (FTY720), a new type of immunosuppressive agent, is approved for the treatment of multiple sclerosis (MS) by United States food and drug administration (FDA) in 2010 [9], [27]. Previously, it has been reported that FTY720 has protective effect on intracerebral hemorrhage-induced secondary neurological damage in rats [24]. Besides, in the rodent model of autism, FTY720 improved short-term memory in 8-arm radial maze test [26], indicating that FTY720 possesses potent neuroprotective activity. FTY720 alleviated the symptoms of ischemia/reperfusion injury in animal model and reduced IL-23 levels in brain tissues [21]. Nevertheless, the underlying mechanism how FTY720 exerts protective effect on brain remains incompletely understood and application of FTY720 in treatment of cerebral ischemia/reperfusion injury would be worth investigating further.

Inflammatory cytokines play crucial roles during cerebral ischemia–reperfusion, especially in the acute phase within 24 h [26], [38]. p38MAPK (mitogen-activated protein kinase, MAPK) signaling pathway was activated in ischemic brain tissue and p38MAPK phosphorylation was implicated in the activation of inflammatory response [12], [20]. Previous work suggested that p38MAPK activity was increased in microglia of the hippocampus, and p38MAPK inhibitors effectively reduced neuronal death in rats undergoing bilateral common carotid artery ligation (BCCAL), followed by reperfusion for 7 min [36], [37]. The purpose of this study is to investigate whether FTY720 improves neurological deficits and reduces neuronal apoptosis in rodent model of middle cerebral ischemia reperfusion, and whether this protective effect is associated with p38MAPK and NF-κB signaling pathways.

In this study, using a rat model of middle cerebral ischemia reperfusion caused by bilateral common carotid artery ligation, we found that FTY720 improved the neurological deficits, reduced neuronal damage and decreased the release of inflammatory cytokines (IL-1β, IL-6, and TNF-α). Given the neuroprotective activity of FTY720, it may be a potential therapeutic agent for patients with ischemia reperfusion injury-associated tissue inflammation and organ dysfunction.

Section snippets

Animals

Sprague-Dawley (SD) rats (250–300 g, N = 76 rats in total, male) were purchased from the Experimental Animal Center of Jinzhou Medical University and housed under specific-pathogen free (SPF) condition with access to food and water ad libitum (temperature 22–24 °C, relative humidity 45–55%, and12:12 h light‐dark cycle). All Animal experiments were approved by the Ethics Committee of Jinzhou Medical University.

Rat model of transient cerebral ischemia/reperfusion (tMCAO/R)

Cerebral ischemia was generated by transient middle cerebral artery occlusion (tMCAO).

FTY720 improves neurological deficits of SD rats undergoing cerebral ischemia reperfusion injury

In present study, experimental workflow is represented in Fig. 1A. We found that neurological score in the operated groups was significantly increased compared with sham-operated groups, suggesting we successfully construct cerebral ischemia reperfusion injury rat model (Fig. 1B, 1C). When rats were administrated with two different doses of FTY720 before tMACO/R, neurological scores in FTY720-pretreatment groups were significantly lower than that in the operated group in a dose-dependent manner

Discussion

In the present study, FTY720 was applied to interfere with the progression of cerebral ischemia reperfusion injury in rats. Based on a rat cerebral ischemia–reperfusion injury model, data showed that the neurological score of rats in the FTY720 pretreatment or post-treatment group was significantly lower than that in the operated group at days 7, indicating that FTY720 could improve neurological symptoms caused by ischemia and reperfusion. Additionally, FTY720 administration before or after

Conclusion

In summary, our findings suggested that FTY720 ameliorated neurological deficit of rat cerebral ischemia reperfusion model and reduced neuronal damage in the ischemic brain., FTY720 treatment down-regulated the expression levels of inflammatory factors including IL-1β, IL-6 and TNF-α, and inhibited the activation of P38 MAPK and NF-κB signaling pathways. FTY720 holds promise as an attractive therapeutic drug for alleviating ischemic perfusion injury.

Authors’ contributions

Ling Zhang, Rubo Sui and Lei Zhang contributed to the animal model performance, collection and assembly of data, and drafting the manuscript. Rubo Sui and Lei Zhang involved in experimental design, interpretation of data, critical revision of the manuscript. All authors read and approved the final manuscript.

Ethics approval

All rats were obtained from the Laboratory Animal Center of Jinzhou Medical University. All animal procedures followed the National Institutes of Health’s Guide for the Care and Use of Laboratory Animals. All protocols were approved on accordance of the Committee on Animal Research of Jinzhou Medical University.

Declaration of Competing Interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Acknowledgments

Not applicable.

Funding

This work was supported by the National Natural Science Foundation of China (Grant No. 81371461), Fund from Liaoning Education Department (Grant No. JYTQN201707), the Natural Science Foundation of Liaoning Province (Grant No. 2019-ZD-0802), and the Natural Science Foundation of Liaoning Provincial Department of Education (Grant No. JYTJCZR201903; Grant No. JYTJCZR2020065).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

References (38)

  • C. Benakis et al.

    The role of microglia and myeloid immune cells in acute cerebral ischemia

    Front. Cell. Neurosci.

    (2014)
  • F. Campos et al.

    Fingolimod reduces hemorrhagic transformation associated with delayed tissue plasminogen activator treatment in a mouse thromboembolic model

    Stroke

    (2013)
  • B.Z. Chaudhry et al.

    Sphingosine 1-Phosphate Receptor Modulators for the Treatment of Multiple Sclerosis

    Neurotherapeutics : the journal of the American Society for Experimental NeuroTherapeutics

    (2017)
  • Y.-L. Cheng et al.

    Evidence that neuronal Notch-1 promotes JNK/c-Jun activation and cell death following ischemic stress

    Brain Res.

    (2014)
  • M. Cieślak et al.

    Relationship between the induction of inflammatory processes and infectious diseases in patients with ischemic stroke

    Acta Biochim. Pol.

    (2013)
  • Y.F. Dong et al.

    S1PR3 is essential for phosphorylated fingolimod to protect astrocytes against oxygen-glucose deprivation-induced neuroinflammation via inhibiting TLR2/4-NFkappaB signalling

    J. Cell Mol. Med.

    (2018)
  • H.C. Emsley et al.

    A randomised phase II study of interleukin-1 receptor antagonist in acute stroke patients

    J. Neurol. Neurosurg. Psychiatry

    (2005)
  • S. Fan et al.

    Thymus quinquecostatus Celak. ameliorates cerebral ischemia-reperfusion injury via dual antioxidant actions: Activating Keap1/Nrf2/HO-1 signaling pathway and directly scavenging ROS

    Phytomedicineinternational journal of phytotherapy and phytopharmacology

    (2021)
  • C.A. Foster et al.

    Brain penetration of the oral immunomodulatory drug FTY720 and its phosphorylation in the central nervous system during experimental autoimmune encephalomyelitis: consequences for mode of action in multiple sclerosis

    The Journal of pharmacology and experimental therapeutics

    (2007)
  • G. Fredman et al.

    Resolvin D1 limits 5-lipoxygenase nuclear localization and leukotriene B4 synthesis by inhibiting a calcium-activated kinase pathway

    PNAS

    (2014)
  • X.D. Guo et al.

    FTY720 Exerts Anti-Glioma Effects by Regulating the Glioma Microenvironment Through Increased CXCR4 Internalization by Glioma-Associated Microglia

    Front. Immunol.

    (2020)
  • Z. Guo et al.

    Lipoxin A4 Reduces Inflammation Through Formyl Peptide Receptor 2/p38 MAPK Signaling Pathway in Subarachnoid Hemorrhage Rats

    Stroke

    (2016)
  • Y.u. Hasegawa et al.

    Activation of sphingosine 1-phosphate receptor-1 by FTY720 is neuroprotective after ischemic stroke in rats

    Stroke

    (2010)
  • R.L. Jayaraj et al.

    Neuroinflammation: friend and foe for ischemic stroke

    Journal of neuroinflammation

    (2019)
  • R. Jin et al.

    Role of inflammation and its mediators in acute ischemic stroke

    Journal of cardiovascular translational research

    (2013)
  • R. Jin et al.

    Inflammatory mechanisms in ischemic stroke: role of inflammatory cells

    J. Leukoc. Biol.

    (2010)
  • Z. Jin et al.

    The effect of stress on stroke recovery in a photothrombotic stroke animal model

    Brain Res.

    (2010)
  • T. Kirino

    Delayed neuronal death in the gerbil hippocampus following ischemia

    Brain Res.

    (1982)
  • T. Li et al.

    Dapk1 improves inflammation, oxidative stress and autophagy in LPS-induced acute lung injury via p38MAPK/NF-kappaB signaling pathway

    Mol. Immunol.

    (2020)
  • Cited by (0)

    View full text